Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
1.
Journal of Veterinary Science ; : 235-244, 2012.
Article in English | WPRIM | ID: wpr-65170

ABSTRACT

Akt/protein kinase B is a well-known cell survival factor and activated by many stimuli including mechanical stretching. Therefore, we evaluated the cardioprotective effect of a brief mechanical stretching of rat hearts and determined whether activation of Akt through phosphatidylinositol 3-kinase (PI3K) is involved in stretch-induced cardioprotection (SIC). Stretch preconditioning reduced infarct size and improved post-ischemic cardiac function compared to the control group. Phosphorylation of Akt and its downstream substrate, GSK-3beta, was increased by mechanical stretching and completely blocked by wortmannin, a PI3K inhibitor. Treatment with lithium or SB216763 (GSK-3beta inhibitors) before ischemia induction mimicked the protective effects of SIC on rat heart. Gadolinium (Gd3+), a blocker of stretch-activated ion channels (SACs), inhibited the stretch-induced phosphorylation of Akt and GSK-3beta. Furthermore, SIC was abrogated by wortmannin and Gd3+. In vivo stretching induced by an aorto-caval shunt increased Akt phosphorylation and reduced myocardial infarction; these effects were diminished by wortmannin and Gd3+ pretreatment. Our results showed that mechanical stretching can provide cardioprotection against ischemia-reperfusion injury. Additionally, the activation of Akt, which might be regulated by SACs and the PI3K pathway, plays an important role in SIC.


Subject(s)
Animals , Male , Rats , Androstadienes/pharmacology , Gadolinium/pharmacology , Glycogen Synthase Kinase 3/metabolism , Indoles/pharmacology , Ischemic Preconditioning, Myocardial , Lithium/pharmacology , Maleimides/pharmacology , Myocardial Reperfusion Injury/enzymology , Phosphatidylinositol 3-Kinase/antagonists & inhibitors , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Random Allocation , Rats, Sprague-Dawley , Specific Pathogen-Free Organisms
2.
Experimental & Molecular Medicine ; : 403-411, 2012.
Article in English | WPRIM | ID: wpr-57559

ABSTRACT

The localization of estrogen (E2) has been clearly shown in hippocampus, called local hippocampal E2. It enhanced neuronal synaptic plasticity and protected neuron form cerebral ischemia, similar to those effects of exogenous E2. However, the interactive function of hippocampal and exogenous E2 on synaptic plasticity activation and neuroprotection is still elusive. By using hippocampal H19-7 cells, we demonstrated the local hippocampal E2 that totally suppressed by aromatase inhibitor anastrozole. Anastrozole also suppressed estrogen receptor (ER)beta, but not ERalpha, expression. Specific agonist of ERalpha (PPT) and ERbeta (DPN) restored ERbeta expression in anastrozole-treated cells. In combinatorial treatment with anastrozole and phosphoinositide kinase-3 (PI-3K) signaling inhibitor wortmannin, PPT could not improve hippocampal ERbeta expression. On the other hand, DPN induced basal ERbeta translocalization into nucleus of anastrozole-treated cells. Exogenous E2 increased synaptic plasticity markers expression in H19-7 cells. However, exogenous E2 could not enhance synaptic plasticity in anastrozole-treated group. Exogenous E2 also increased cell viability and B-cell lymphoma 2 (Bcl2) expression in H2O2-treated cells. In combined treatment of anastrozole and H2O2, exogenous E2 failed to enhance cell viability and Bcl2 expression in hippocampal H19-7 cells. Our results provided the evidence of the priming role of local hippocampal E2 on exogenous E2-enhanced synaptic plasticity and viability of hippocampal neurons.


Subject(s)
Animals , Rats , Androstadienes/pharmacology , Aromatase Inhibitors/pharmacology , Cell Line , Cell Survival/drug effects , Estrogen Receptor alpha/agonists , Estrogen Receptor beta/agonists , Estrogens/metabolism , Hippocampus/cytology , Hydrogen Peroxide/pharmacology , Nervous System/drug effects , Neuronal Plasticity/drug effects , Neuroprotective Agents , Nitriles/pharmacology , Phosphatidylinositol 3-Kinase/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Triazoles/pharmacology
3.
Experimental & Molecular Medicine ; : 153-161, 2006.
Article in English | WPRIM | ID: wpr-15696

ABSTRACT

Adhesion and migration of vascular smooth muscle cells (VSMCs) play an important role in the pathogenesis of atherosclerosis. These processes involve the interaction of VSMCs with extracellular matrix proteins. Here, we investigated integrin isoforms and signaling pathways mediating the adhesion and migration of VSMCs on betaig-h3, a transforming growth factor (TGF)-beta-inducible extracellular matrix protein that is elevated in atherosclerotic plaques. Adhesion assays showed that the alphavbeta5 integrin is a functional receptor for the adhesion of aortic VSMCs to betaig-h3. An YH18 motif containing amino acids between 563 and 580 of betaig-h3 was an essential motif for the adhesion and growth of VSMCs. Interaction between the YH18 motif and the alphavbeta5 integrin was responsible for the migration of VSMCs on betaig-h3. Inhibitors of phosphatidylinositide 3-kinase, extracellular signal-regulated kinase (ERK), and Src kinase reduced the adhesion and migration of VSMCs on betaig-h3. betaig-h3 triggered phosphorylation and activation of AKT, ERK, focal adhesion kinase, and paxillin mediating the adhesion and migration of VSMCs. Taken together, these results suggest that betaig-h3 and alphavbeta5 integrin play a role in the adhesion and migration of VSMCs during the pathogenesis of atherosclerosis.


Subject(s)
Humans , Animals , src-Family Kinases/antagonists & inhibitors , Transforming Growth Factor beta/genetics , Signal Transduction/physiology , Receptors, Vitronectin/genetics , Protein-Tyrosine Kinases/antagonists & inhibitors , Paxillin/metabolism , Myocytes, Smooth Muscle/drug effects , Muscle, Smooth, Vascular/cytology , Morpholines/pharmacology , Molecular Sequence Data , Integrins/genetics , Flavonoids/pharmacology , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Matrix Proteins/genetics , Enzyme Inhibitors/pharmacology , Chromones/pharmacology , Cells, Cultured , Cell Movement/physiology , Cell Adhesion/physiology , Amino Acid Sequence , Amino Acid Motifs/genetics , Phosphatidylinositol 3-Kinase/antagonists & inhibitors
4.
Experimental & Molecular Medicine ; : 588-600, 2005.
Article in English | WPRIM | ID: wpr-191492

ABSTRACT

Prostaglandin E2(PGE2), a major product of cyclooxygenase, has been implicated in modulating angiogenesis, vascular function, and inflammatory processes, but the underlying mechanism is not clearly elucidated. We here investigated the molecular mechanism by which PGE 2 regulates angiogenesis. Treatment of human umbilical vein endothelial cells (HUVEC) with PGE 2 increased angiogenesis. PGE 2 increased phosphorylation of Akt and endothelial nitric oxide synthase (eNOS), eNOS activity, and nitric oxide (NO) production by the activation of cAMP-dependent protein kinase (PKA) and phosphatidylinositol 3-kinase (PI3K). Dibutyryl cAMP (DB-cAMP) mimicked the role of PGE 2 in angiogenesis and the signaling pathway, suggesting that cAMP is a down-stream mediator of PGE 2. Furthermore, PGE 2 increased endothelial cell sprouting from normal murine aortic segments, but not from eNOS-deficient ones, on Matrigel. The angiogenic effects of PGE 2 were inhibited by the inhibitors of PKA, PI3K, eNOS, and soluble guanylate cyclase, but not by phospholipase C inhibitor. These results clearly show that PGE 2 increased angiogenesis by activating the NO/cGMP signaling pathway through PKA/PI3K/Akt-dependent increase in eNOS activity.


Subject(s)
Animals , Humans , Mice , Rats , Phosphatidylinositol 3-Kinase/antagonists & inhibitors , Aorta , Cell Movement/drug effects , Cell Proliferation/drug effects , Cyclic AMP/metabolism , Cyclic GMP/biosynthesis , Dinoprostone/pharmacology , Endothelial Cells/drug effects , Enzyme Inhibitors/pharmacology , Mice, Knockout , Neovascularization, Physiologic/drug effects , Nitric Oxide/biosynthesis , Nitric Oxide Synthase Type III/deficiency , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Rats, Sprague-Dawley , Signal Transduction/drug effects , Umbilical Veins/cytology
5.
Journal of Korean Medical Science ; : 161-167, 2002.
Article in English | WPRIM | ID: wpr-197897

ABSTRACT

Nitric oxide (NO) seems to play a pivotal role in the vascular endothelial growth factor (VEGF)-induced endothelial cell proliferation. This study was designed to investigate the role and intracellular signal pathway of endothelial nitric oxide synthase (eNOS) activation induced by VEGF. ECV 304 cells were treated with betaVEGF(165) and then cell proliferation, eNOS protein and mRNA expression levels were analyzed to elucidate the functional role of eNOS in cell proliferation induced by VEGF. After exposure of cells to betaVEGF(165) , eNOS activity and cell growth were increased by approximately two-fold in the betaVEGF(165) -treated cells compared to the untreated cells. In addition, VEGF stimulated eNOS expression at both the mRNA and protein levels in a dose-dependent manner. Phosphatidylinositol-3 kinase (PI-3K) inhibitors were used to assess PI-3K involvement in eNOS regulation. LY294002 was found to attenuate VEGF-stimulated eNOS expression. Wortmannin was not as effective as LY294002, but the reduction effect was detectable. Cells activated by VEGF showed increased ERK1/2 levels. Moreover, the VEGF-induced eNOS expression was reduced by the PD98059, MAPK pathway inhibitor. This suggests that eNOS expression might be regulated by PI-3K and the ERK1/2 signaling pathway. In conclusion, betaVEGF(165) induces ECV 304 cell proliferation via the NO produced by eNOS. In addition, eNOS may be regulated by the PI-3K or mitogen-activated protein kinase pathway.


Subject(s)
Phosphatidylinositol 3-Kinase/antagonists & inhibitors , Cell Division/drug effects , Cell Line , Endothelial Growth Factors/metabolism , Endothelium, Vascular/cytology , Gene Expression Regulation, Enzymologic , Lymphokines/metabolism , MAP Kinase Signaling System , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type III , Signal Transduction , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
SELECTION OF CITATIONS
SEARCH DETAIL